Sign in to use this feature.

Years

Between: -

Subjects

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Journals

Article Types

Countries / Regions

Search Results (69)

Search Parameters:
Keywords = microtubule depolymerization

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
34 pages, 6455 KiB  
Article
IBCar: Potent Orally Bioavailable Methyl N-[5-(3′-Iodobenzoyl)-1H-Benzimidazol-2-yl]Carbamate for Breast Cancer Therapy
by Janina Baranowska-Kortylewicz and Ying Yan
Cancers 2025, 17(15), 2526; https://doi.org/10.3390/cancers17152526 - 30 Jul 2025
Viewed by 220
Abstract
Objectives: To investigate the efficacy and underlying mechanisms of IBCar’s biological activity in breast cancer models, both in cell culture and in mice, and to compare its effects on cancer versus normal cells. Methods: The cytotoxicity of IBCar was evaluated using [...] Read more.
Objectives: To investigate the efficacy and underlying mechanisms of IBCar’s biological activity in breast cancer models, both in cell culture and in mice, and to compare its effects on cancer versus normal cells. Methods: The cytotoxicity of IBCar was evaluated using the MTS assay to assess metabolic activity and the clonogenic assay to determine reproductive integrity. The impact of IBCar on microtubule integrity, mitochondrial function, and multiple signaling pathways was analyzed using Western blotting, microarray analysis, and live cell imaging. The therapeutic effectiveness of orally administered IBCar was assessed in a transgenic mouse model of Luminal B breast cancer and in mice implanted with subcutaneous triple-negative breast cancer xenografts. Results: IBCar demonstrated potent cytotoxicity across a diverse panel of breast cancer cell lines, including those with mutant or wild-type TP53, and cell lines with short and long doubling times. Comparative analysis revealed distinct responses between normal and cancer cells, including differences in IBCar’s effects on the mitochondrial membrane potential, endoplasmic reticulum stress and activation of cell death pathways. In breast cancer cells, IBCar was cytotoxic at nanomolar concentrations, caused irreversible microtubule depolymerization leading to sustained mitochondrial dysfunction, endoplasmic reticulum stress, and induced apoptosis. In normal cells, protective mechanisms included reversible microtubule depolymerization and activation of pro-survival signaling via the caspase-8 and riptosome pathways. The therapeutic potential of IBCar was confirmed in mouse models of Luminal B and triple negative BC, where it exhibited strong antitumor activity without detectable toxicity. Conclusions: These findings collectively support IBCar as a promising, effective, and safe therapeutic candidate for breast cancer treatment. Full article
Show Figures

Figure 1

24 pages, 2279 KiB  
Article
Insights into the Structural Patterns in Human Glioblastoma Cell Line SF268 Activity and ADMET Prediction of Curcumin Derivatives
by Lorena Coronado, Johant Lakey-Beitia, Marisin Pecchio, Michelle G. Ng, Ricardo Correa, Gerardo Samudio-Ríos, Jessica Cruz-Mora, Arelys L. Fuentes, K. S. Jagannatha Rao and Carmenza Spadafora
Pharmaceutics 2025, 17(8), 968; https://doi.org/10.3390/pharmaceutics17080968 - 25 Jul 2025
Viewed by 340
Abstract
Background/Objectives: Curcumin is a promising therapy for glioblastoma but is limited by poor water solubility, rapid metabolism, and low blood–brain barrier penetration. This study aimed to evaluate curcumin and six curcumin derivatives with improved activity against a glioblastoma cell line and favorable [...] Read more.
Background/Objectives: Curcumin is a promising therapy for glioblastoma but is limited by poor water solubility, rapid metabolism, and low blood–brain barrier penetration. This study aimed to evaluate curcumin and six curcumin derivatives with improved activity against a glioblastoma cell line and favorable absorption, distribution, metabolism, excretion, and toxicity (ADMET) properties. Methods: Twenty-one curcumin derivatives were assessed and subjected to in vitro MTT cytotoxicity assays in SF268 glioblastoma and Vero cells. On the basis of the cytotoxicity results, six derivatives with the most favorable characteristics were selected for additional mechanistic studies, which included microtubule depolymerization, mitochondrial membrane potential (ΔΨm), and BAX activation assays. ADMET properties were determined in silico. Results: Compounds 24, 6, and 11 demonstrated better activity (IC50: 0.59–3.97 µg/mL and SI: 3–20) than curcumin (IC50: 6.3 µg/mL; SI: 2.5). Lead derivatives destabilized microtubules, induced ΔΨm collapse, and activated BAX. In silico ADMET prediction analysis revealed that compounds 4 and 6 were the most promising for oral administration from a biopharmaceutical and pharmacokinetic point of view. Conclusions: Strategic modifications were made to one or both hydroxyl groups of the aromatic rings of curcumin to increase its physicochemical stability and activity against glioblastoma cell line SF268. Compound 4, bearing fully protected aromatic domains, was identified as a prime candidate for in vivo validation and formulation development. Full article
Show Figures

Graphical abstract

15 pages, 2357 KiB  
Article
Development of a Novel, Highly Sensitive System for Evaluating Ebola Virus Particle Formation
by Wakako Furuyama, Miako Sakaguchi, Hanako Ariyoshi and Asuka Nanbo
Viruses 2025, 17(7), 1016; https://doi.org/10.3390/v17071016 - 19 Jul 2025
Viewed by 426
Abstract
Ebola virus (EBOV) causes severe hemorrhagic fevers in humans, and effective countermeasures remain limited. The EBOV-encoded major matrix protein VP40 is essential for viral assembly, budding, and particle release, making it a promising target for antiviral drug development. However, no approved drugs currently [...] Read more.
Ebola virus (EBOV) causes severe hemorrhagic fevers in humans, and effective countermeasures remain limited. The EBOV-encoded major matrix protein VP40 is essential for viral assembly, budding, and particle release, making it a promising target for antiviral drug development. However, no approved drugs currently target the viral particle formation process. In this study, we established a simple and highly sensitive screening system to evaluate VP40-mediated virus-like particle (VLP) formation under biosafety level −2 conditions. The system uses the HiBiT luminescence-based reporter fused to VP40, allowing for the detection of VP40 release. Our results demonstrate that the HiBiT sequence fused at the N-terminus [HiBiT-VP40 (N)] retains VP40′s ability to form VLPs, supporting its use as a functional reporter. Furthermore, we validated the system by assessing the role of Rab11-dependent trafficking in VP40-mediated budding and by evaluating the effect of nocodazole, a microtubule depolymerizer, on VLP release. This novel screening system provides a convenient and reliable platform for screening potential inhibitors targeting the late stages of EBOV infection, including viral particle formation and release. Additionally, its potential adaptability to other filoviruses suggests wide applicability in the discovery and development of additional novel therapeutic agents. Full article
Show Figures

Figure 1

37 pages, 4217 KiB  
Article
A Benzodiazepine-Derived Molecule That Interferes with the Bio-Mechanical Properties of Glioblastoma-Astrocytoma Cells Altering Their Proliferation and Migration
by Gregorio Ragazzini, Andrea Mescola, Riccardo Tassinari, Alessia Gallerani, Chiara Zannini, Domenico Di Rosa, Claudia Cavallini, Martina Marcuzzi, Valentina Taglioli, Beatrice Bighi, Roberta Ettari, Vincenzo Zappavigna, Carlo Ventura, Andrea Alessandrini and Lorenzo Corsi
Int. J. Mol. Sci. 2025, 26(6), 2767; https://doi.org/10.3390/ijms26062767 - 19 Mar 2025
Cited by 2 | Viewed by 794
Abstract
Glioblastoma multiforme (grade IV glioma) is characterized by a high invasive potential, making surgical intervention extremely challenging and patient survival very limited. Current pharmacological approaches show, at best, slight improvements in the therapy against this type of tumor. Microtubules are often the target [...] Read more.
Glioblastoma multiforme (grade IV glioma) is characterized by a high invasive potential, making surgical intervention extremely challenging and patient survival very limited. Current pharmacological approaches show, at best, slight improvements in the therapy against this type of tumor. Microtubules are often the target of antitumoral drugs, and specific drugs affecting their dynamics by acting on microtubule-associated proteins (MAPs) without producing their depolymerization could affect both glioma cell migration/invasion and cell proliferation. Here, we analyzed on a cellular model of glioblastoma multiforme, the effect of a molecule (1-(4-amino-3,5-dimethylphenyl)-3,5-dihydro-7,8-ethylenedioxy-4h2,3-benzodiazepin-4-one, hereafter named 1g) which was shown to act as a cytostatic drug in other cell types by affecting microtubule dynamics. We found that the molecule acts also as a migration suppressor by inducing a loss of cell polarity. We characterized the mechanics of U87MG cell aggregates exposed to 1g by different biophysical techniques. We considered both 3D aggregates and 2D cell cultures, testing substrates of different stiffness. We established that this molecule produces a decrease of cell spheroid contractility and it impairs 3D cell invasion. At the same time, in the case of isolated cells, 1g selectively produces an almost instantaneous loss of cell polarity blocking migration and it also produces a disorganization of the mitotic spindle when cells reach mitosis, leading to frequent mitotic slippage events followed by cell death. We can state that the studied molecule produces similar effects to other molecules that are known to affect the dynamics of microtubules, but probably indirectly via microtubule-associated proteins (MAPs) and following different biochemical pathways. Consistently, we report evidence that, regarding its effect on cell morphology, this molecule shows a specificity for some cell types such as glioma cells. Interestingly, being a molecule derived from a benzodiazepine, the 1g chemical structure could allow this molecule to easily cross the blood–brain barrier. Thanks to its chemical/physical properties, the studied molecule could be a promising new drug for the specific treatment of GBM. Full article
(This article belongs to the Special Issue Biomechanics and Molecular Research on Glioblastoma)
Show Figures

Figure 1

20 pages, 5298 KiB  
Article
SNX19 Interacts with Caveolin-1 and Flotillin-1 to Regulate D1R Endocytosis and Signaling
by Bibhas Amatya, Jacob Q. M. Polzin, Van A. M. Villar, Jiang Yang, Prasad Konkalmatt, Xiaoyan Wang, Raisha C. Cadme, Peng Xu, John J. Gildea, Santiago Cuevas, Ines Armando, Robin A. Felder, Pedro A. Jose and Hewang Lee
Biomedicines 2025, 13(2), 481; https://doi.org/10.3390/biomedicines13020481 - 15 Feb 2025
Viewed by 907
Abstract
Background: Sorting nexin 19 (SNX19) is important in the localization and trafficking of the dopamine D1 receptor (D1R) to lipid raft microdomains. However, the interaction between SNX19 and the lipid raft components caveolin-1 or flotillin-1 and, in particular, their roles [...] Read more.
Background: Sorting nexin 19 (SNX19) is important in the localization and trafficking of the dopamine D1 receptor (D1R) to lipid raft microdomains. However, the interaction between SNX19 and the lipid raft components caveolin-1 or flotillin-1 and, in particular, their roles in the cellular endocytosis and cell membrane trafficking of the D1R have not been determined. Methods: Caveolin-1 and flotillin-1 motifs were analyzed by in silico analysis; colocalization was observed by confocal immunofluorescence microscopy; protein-protein interaction was determined by co-immunoprecipitation. Results: In silico analysis revealed the presence of putative caveolin-1 and flotillin-1 binding motifs within SNX19. In mouse and human renal proximal tubule cells (RPTCs), SNX19 was localized mainly in lipid rafts. In mouse RPTCs transfected with wild-type (WT) Snx19, fenoldopam (FEN), a D1-like receptor agonist, increased the colocalization of SNX19 with caveolin-1 and flotillin-1. FEN also increased the co-immunoprecipitation of SNX19 with caveolin-1 and flotillin-1, effects that were prevented by SCH39166, a D1-like receptor antagonist. The FEN-mediated increase in the residence of SNX19 in lipid rafts and the colocalization of the D1R with caveolin-1 and flotilin-1 were attenuated by the deletion of a caveolin-1 (YHTVNRRYREF) (ΔCav1) or a flotillin-1 (EEGPGTETETGLPVS) (ΔFlot1) binding motif. The FEN-mediated increase in intracellular cAMP production was also impaired by the deletion of either the flotillin-1 or caveolin-1 binding motif. Nocodazole, a microtubule depolymerization inhibitor, interfered with the FEN-mediated increase in the colocalization between SNX19 and D1R. Conclusion: SNX19 contains caveolin-1 and flotillin-1 binding motifs, which play an important role in D1R endocytosis and signaling. Full article
(This article belongs to the Special Issue Dopamine Signaling Pathway in Health and Disease—2nd Edition)
Show Figures

Graphical abstract

22 pages, 2306 KiB  
Review
From Deworming to Cancer Therapy: Benzimidazoles in Hematological Malignancies
by Upendarrao Golla, Satyam Patel, Nyah Shah, Stella Talamo, Riya Bhalodia, David Claxton, Sinisa Dovat and Arati Sharma
Cancers 2024, 16(20), 3454; https://doi.org/10.3390/cancers16203454 - 12 Oct 2024
Cited by 2 | Viewed by 5949
Abstract
Drug repurposing is a strategy to discover new therapeutic uses for existing drugs, which have well-established toxicity profiles and are often more affordable. This approach has gained significant attention in recent years due to the high costs and low success rates associated with [...] Read more.
Drug repurposing is a strategy to discover new therapeutic uses for existing drugs, which have well-established toxicity profiles and are often more affordable. This approach has gained significant attention in recent years due to the high costs and low success rates associated with traditional drug development. Drug repositioning offers a more time- and cost-effective path for identifying new treatments. Several FDA-approved non-chemotherapy drugs have been investigated for their anticancer potential. Among these, anthelmintic benzimidazoles (such as albendazole, mebendazole, and flubendazole) have garnered interest due to their effects on microtubules and oncogenic signaling pathways. Blood cancers, which frequently develop resistance and have high mortality rates, present a critical need for effective therapies. This review highlights the recent advances in repurposing benzimidazoles for blood malignancies. These compounds induce cell cycle arrest, differentiation, tubulin depolymerization, loss of heterozygosity, proteasomal degradation, and inhibit oncogenic signaling to exert their anticancer effects. We also discuss current limitations and strategies to overcome them, emphasizing the potential of combining benzimidazoles with standard therapies for improved treatment of hematological cancers. Full article
(This article belongs to the Special Issue Drug Repurposing and Reformulation for Cancer Treatment: 2nd Edition)
Show Figures

Graphical abstract

24 pages, 9864 KiB  
Article
Synthesis and Antiproliferative Effect of 3,4,5-Trimethoxylated Chalcones on Colorectal and Prostatic Cancer Cells
by Cécile Letulle, François-Xavier Toublet, Aline Pinon, Soufyane Hba, Aurélie Laurent, Vincent Sol, Catherine Fagnère, Benjamin Rioux, Florent Allais, Sophie Michallet, Laurence Lafanechère, Youness Limami, Mounia Oudghiri, Mohamed Othman, Adam Daïch, Bertrand Liagre, Ata Martin Lawson and Christelle Pouget
Pharmaceuticals 2024, 17(9), 1207; https://doi.org/10.3390/ph17091207 - 13 Sep 2024
Cited by 2 | Viewed by 1770
Abstract
In the context of designing innovative anticancer agents, the synthesis of a series of chalcones bearing a 3,4,5-trimethoxylated A ring and a variety of B rings, including phenols and original heterocycles such as chromones, was conducted. For this end, Claisen–Schmidt condensation was performed [...] Read more.
In the context of designing innovative anticancer agents, the synthesis of a series of chalcones bearing a 3,4,5-trimethoxylated A ring and a variety of B rings, including phenols and original heterocycles such as chromones, was conducted. For this end, Claisen–Schmidt condensation was performed in basic or acidic conditions between the common starting material 3,4,5-trimethoxyacetophenone and appropriate aldehydes; this allowed the recovery of fifteen chalcones in moderate–good yields. The synthesized compounds were screened for their antiproliferative activity against colorectal and prostatic cancer cells, using a colorimetric MTT assay. Among the new chromonyl series, chalcone 13 demonstrates an interesting antiproliferative effect, with IC50 values in the range of 2.6–5.1 µM at 48 h. Then, our study evidenced that indolyl chalcone 10 exhibits excellent activity towards the selected cell lines (with IC50 less than 50 nM). This compound has already been described and has been shown to be a potent anticancer agent against other cancer cell lines. Our investigations highlighted apoptosis induction, through several pro-apoptotic markers, of these two heterocyclic chalcones. Considering phenolic chalcones, compounds 2 and 8 were found to be the most active against cell proliferation, exerting their effect by inducing the depolymerization of cell microtubules. The most promising compounds in this series will be selected for application in a strategy of vectorization by either active or passive targeting. Full article
Show Figures

Figure 1

16 pages, 6851 KiB  
Article
Genome-Wide Characterization of IQD Family Proteins in Apple and Functional Analysis of the Microtubule-Regulating Abilities of MdIQD17 and MdIQD28 under Cold Stress
by Yu Zhang, Shengjie Wang, Chaochao Zhang, Meng Qi, Luoqi Liu, Lipeng Yang and Na Lian
Plants 2024, 13(17), 2532; https://doi.org/10.3390/plants13172532 - 9 Sep 2024
Cited by 1 | Viewed by 1125
Abstract
Microtubules undergo dynamic remodeling in response to diverse abiotic stress in plants. The plant-specific IQ67 DOMAIN (IQD) family proteins serve as microtubule-associated proteins, playing multifaceted roles in plant development and response to abiotic stress. However, the biological function of IQD genes in apple [...] Read more.
Microtubules undergo dynamic remodeling in response to diverse abiotic stress in plants. The plant-specific IQ67 DOMAIN (IQD) family proteins serve as microtubule-associated proteins, playing multifaceted roles in plant development and response to abiotic stress. However, the biological function of IQD genes in apple remains unclear. In this study, we conducted a comprehensive analysis of the Malus domestica genome, identifying 42 IQD genes distributed across 17 chromosomes and categorized them into four subgroups. Promoter analysis revealed the presence of stress-responsive elements. Subsequent expression analysis highlighted the significant upregulation of MdIQD17 and MdIQD28 in response to cold treatments, prompting their selection for further functional investigation. Subcellular localization studies confirmed the association of MdIQD17 and MdIQD28 with microtubules. Crucially, confocal microscopy and quantification revealed diminished microtubule depolymerization in cells transiently overexpressing MdIQD17 and MdIQD28 compared to wild-type cells during cold conditions. In conclusion, this study provides a comprehensive analysis of IQD genes in apple, elucidating their molecular mechanism in response to cold stress. Full article
(This article belongs to the Special Issue Advances in Plant Anatomy and Cell Biology)
Show Figures

Figure 1

20 pages, 4298 KiB  
Article
Centrosomal Protein 55 Regulates Chromosomal Instability in Cancer Cells by Controlling Microtubule Dynamics
by Stefanie Muhs, Themistoklis Paraschiakos, Paula Schäfer, Simon A. Joosse and Sabine Windhorst
Cells 2024, 13(16), 1382; https://doi.org/10.3390/cells13161382 - 20 Aug 2024
Cited by 2 | Viewed by 1600
Abstract
Centrosomal Protein 55 (CEP55) exhibits various oncogenic activities; it regulates the PI3K-Akt-pathway, midbody abscission, and chromosomal instability (CIN) in cancer cells. Here, we analyzed the mechanism of how CEP55 controls CIN in ovarian and breast cancer (OvCa) cells. Down-regulation of CEP55 reduced CIN [...] Read more.
Centrosomal Protein 55 (CEP55) exhibits various oncogenic activities; it regulates the PI3K-Akt-pathway, midbody abscission, and chromosomal instability (CIN) in cancer cells. Here, we analyzed the mechanism of how CEP55 controls CIN in ovarian and breast cancer (OvCa) cells. Down-regulation of CEP55 reduced CIN in all cell lines analyzed, and CEP55 depletion decreased spindle microtubule (MT)-stability in OvCa cells. Moreover, recombinant CEP55 accelerated MT-polymerization and attenuated cold-induced MT-depolymerization. To analyze a potential relationship between CEP55-controlled CIN and its impact on MT-stability, we identified the CEP55 MT-binding peptides inside the CEP55 protein. Thereafter, a mutant with deficient MT-binding activity was re-expressed in CEP55-depleted OvCa cells and we could show that this mutant did not restore reduced CIN in CEP55-depleted cells. This finding strongly indicates that CEP55 regulates CIN by controlling MT dynamics. Full article
(This article belongs to the Section Cell Microenvironment)
Show Figures

Graphical abstract

13 pages, 1000 KiB  
Review
Clostridioides difficile Toxins: Host Cell Interactions and Their Role in Disease Pathogenesis
by Md Zahidul Alam and Rajat Madan
Toxins 2024, 16(6), 241; https://doi.org/10.3390/toxins16060241 - 24 May 2024
Cited by 14 | Viewed by 4658
Abstract
Clostridioides difficile, a Gram-positive anaerobic bacterium, is the leading cause of hospital-acquired antibiotic-associated diarrhea worldwide. The severity of C. difficile infection (CDI) varies, ranging from mild diarrhea to life-threatening conditions such as pseudomembranous colitis and toxic megacolon. Central to the pathogenesis of [...] Read more.
Clostridioides difficile, a Gram-positive anaerobic bacterium, is the leading cause of hospital-acquired antibiotic-associated diarrhea worldwide. The severity of C. difficile infection (CDI) varies, ranging from mild diarrhea to life-threatening conditions such as pseudomembranous colitis and toxic megacolon. Central to the pathogenesis of the infection are toxins produced by C. difficile, with toxin A (TcdA) and toxin B (TcdB) as the main virulence factors. Additionally, some strains produce a third toxin known as C. difficile transferase (CDT). Toxins damage the colonic epithelium, initiating a cascade of cellular events that lead to inflammation, fluid secretion, and further tissue damage within the colon. Mechanistically, the toxins bind to cell surface receptors, internalize, and then inactivate GTPase proteins, disrupting the organization of the cytoskeleton and affecting various Rho-dependent cellular processes. This results in a loss of epithelial barrier functions and the induction of cell death. The third toxin, CDT, however, functions as a binary actin-ADP-ribosylating toxin, causing actin depolymerization and inducing the formation of microtubule-based protrusions. In this review, we summarize our current understanding of the interaction between C. difficile toxins and host cells, elucidating the functional consequences of their actions. Furthermore, we will outline how this knowledge forms the basis for developing innovative, toxin-based strategies for treating and preventing CDI. Full article
Show Figures

Figure 1

16 pages, 5743 KiB  
Article
Enrichment of Deleterious Mutated Genes Involved in Ciliary Function and Histone Modification in Brain Cancer Patient-Derived Xenograft Models
by Hyeongsun Jeong, Hyo Eun Moon, Seongmin Yun, Seung Woo Cho, Hye Ran Park, Sung-Hye Park, Kyungjae Myung, Taejoon Kwon and Sun Ha Paek
Biomedicines 2023, 11(11), 2934; https://doi.org/10.3390/biomedicines11112934 - 30 Oct 2023
Cited by 1 | Viewed by 1767
Abstract
Patient-derived xenograft (PDX) models, which can retain the characteristics of original tumors in an in vivo-mimicking environment, have been developed to identify better treatment options. However, although original tumors and xenograft tissues mostly share oncogenic mutations and global gene expression patterns, their detailed [...] Read more.
Patient-derived xenograft (PDX) models, which can retain the characteristics of original tumors in an in vivo-mimicking environment, have been developed to identify better treatment options. However, although original tumors and xenograft tissues mostly share oncogenic mutations and global gene expression patterns, their detailed mutation profiles occasionally do not overlap, indicating that selection occurs in the xenograft environment. To understand this mutational alteration in xenografts, we established 13 PDX models derived from 11 brain tumor patients and confirmed their histopathological similarity. Surprisingly, only a limited number of somatic mutations were shared between the original tumor and xenograft tissue. By analyzing deleteriously mutated genes in tumors and xenografts, we found that previously reported brain tumor-related genes were enriched in PDX samples, demonstrating that xenografts are a valuable platform for studying brain tumors. Furthermore, mutated genes involved in cilium movement, microtubule depolymerization, and histone methylation were enriched in PDX samples compared with the original tumors. Even with the limitations of the heterogeneity of clinical lesions with a heterotropic model, our study demonstrates that PDX models can provide more information in genetic analysis using samples with high heterogeneity, such as brain tumors. Full article
(This article belongs to the Section Neurobiology and Clinical Neuroscience)
Show Figures

Figure 1

29 pages, 4432 KiB  
Article
The Microtubule-Targeting Agent Pretubulysin Impairs the Inflammatory Response in Endothelial Cells by a JNK-Dependent Deregulation of the Histone Acetyltransferase Brd4
by Tobias F. Primke, Rebecca Ingelfinger, Mohammed A. F. Elewa, Igor Macinkovic, Andreas Weigert, Matthias P. Fabritius, Christoph A. Reichel, Angelika Ullrich, Uli Kazmaier, Luisa D. Burgers and Robert Fürst
Cells 2023, 12(16), 2112; https://doi.org/10.3390/cells12162112 - 21 Aug 2023
Cited by 2 | Viewed by 2113
Abstract
The anti-inflammatory effects of depolymerizing microtubule-targeting agents on leukocytes are known for a long time, but the potential involvement of the vascular endothelium and the underlying mechanistic basis is still largely unclear. Using the recently synthesized depolymerizing microtubule-targeting agent pretubulysin, we investigated the [...] Read more.
The anti-inflammatory effects of depolymerizing microtubule-targeting agents on leukocytes are known for a long time, but the potential involvement of the vascular endothelium and the underlying mechanistic basis is still largely unclear. Using the recently synthesized depolymerizing microtubule-targeting agent pretubulysin, we investigated the anti-inflammatory potential of pretubulysin and other microtubule-targeting agents with respect to the TNF-induced leukocyte adhesion cascade in endothelial cells, to improve our understanding of the underlying biomolecular background. We found that treatment with pretubulysin reduces inflammation in vivo and in vitro via inhibition of the TNF-induced adhesion of leukocytes to the vascular endothelium by down-regulation of the pro-inflammatory cell adhesion molecules ICAM-1 and VCAM-1 in a JNK-dependent manner. The underlying mechanism includes JNK-induced deregulation and degradation of the histone acetyltransferase Bromodomain-containing protein 4. This study shows that depolymerizing microtubule-targeting agents, in addition to their established effects on leukocytes, also significantly decrease the inflammatory activation of vascular endothelial cells. These effects are not based on altered pro-inflammatory signaling cascades, but require deregulation of the capability of cells to enter constructive transcription for some genes, setting a baseline for further research on the prominent anti-inflammatory effects of depolymerizing microtubule-targeting agents. Full article
Show Figures

Figure 1

26 pages, 3615 KiB  
Article
MCAK Inhibitors Induce Aneuploidy in Triple-Negative Breast Cancer Models
by John C. Smith, Stefan Husted, Jay Pilrose, Stephanie C. Ems-McClung, Jane R. Stout, Richard L. Carpenter and Claire E. Walczak
Cancers 2023, 15(13), 3309; https://doi.org/10.3390/cancers15133309 - 23 Jun 2023
Cited by 6 | Viewed by 3347 | Correction
Abstract
Standard of care for triple-negative breast cancer (TNBC) involves the use of microtubule poisons such as paclitaxel, which are proposed to work by inducing lethal levels of aneuploidy in tumor cells. While these drugs are initially effective in treating cancer, dose-limiting peripheral neuropathies [...] Read more.
Standard of care for triple-negative breast cancer (TNBC) involves the use of microtubule poisons such as paclitaxel, which are proposed to work by inducing lethal levels of aneuploidy in tumor cells. While these drugs are initially effective in treating cancer, dose-limiting peripheral neuropathies are common. Unfortunately, patients often relapse with drug-resistant tumors. Identifying agents against targets that limit aneuploidy may be a valuable approach for therapeutic development. One potential target is the microtubule depolymerizing kinesin, MCAK, which limits aneuploidy by regulating microtubule dynamics during mitosis. Using publicly available datasets, we found that MCAK is upregulated in triple-negative breast cancer and is associated with poorer prognoses. Knockdown of MCAK in tumor-derived cell lines caused a two- to five-fold reduction in the IC50 for paclitaxel, without affecting normal cells. Using FRET and image-based assays, we screened compounds from the ChemBridge 50 k library and discovered three putative MCAK inhibitors. These compounds reproduced the aneuploidy-inducing phenotype of MCAK loss, reduced clonogenic survival of TNBC cells regardless of taxane-resistance, and the most potent of the three, C4, sensitized TNBC cells to paclitaxel. Collectively, our work shows promise that MCAK may serve as both a biomarker of prognosis and as a therapeutic target. Full article
(This article belongs to the Special Issue The Role of the Cytoskeleton in Tumor Progression)
Show Figures

Figure 1

18 pages, 4180 KiB  
Article
Systematic Studies on Anti-Cancer Evaluation of Stilbene and Dibenzo[b,f]oxepine Derivatives
by Filip Borys, Piotr Tobiasz, Marcin Poterała, Hanna Fabczak, Hanna Krawczyk and Ewa Joachimiak
Molecules 2023, 28(8), 3558; https://doi.org/10.3390/molecules28083558 - 18 Apr 2023
Cited by 8 | Viewed by 2512
Abstract
Cancer is one of the most common causes of human death worldwide; thus, numerous therapies, including chemotherapy, have been and are being continuously developed. In cancer cells, an aberrant mitotic spindle—a microtubule-based structure necessary for the equal splitting of genetic material between daughter [...] Read more.
Cancer is one of the most common causes of human death worldwide; thus, numerous therapies, including chemotherapy, have been and are being continuously developed. In cancer cells, an aberrant mitotic spindle—a microtubule-based structure necessary for the equal splitting of genetic material between daughter cells—leads to genetic instability, one of the hallmarks of cancer. Thus, the building block of microtubules, tubulin, which is a heterodimer formed from α- and β-tubulin proteins, is a useful target in anti-cancer research. The surface of tubulin forms several pockets, i.e., sites that can bind factors that affect microtubules’ stability. Colchicine pockets accommodate agents that induce microtubule depolymerization and, in contrast to factors that bind to other tubulin pockets, overcome multi-drug resistance. Therefore, colchicine-pocket-binding agents are of interest as anti-cancer drugs. Among the various colchicine-site-binding compounds, stilbenoids and their derivatives have been extensively studied. Herein, we report systematic studies on the antiproliferative activity of selected stilbenes and oxepine derivatives against two cancer cell lines—HCT116 and MCF-7—and two normal cell lines—HEK293 and HDF-A. The results of molecular modeling, antiproliferative activity, and immunofluorescence analyses revealed that compounds 1a, 1c, 1d, 1i, 2i, 2j, and 3h were the most cytotoxic and acted by interacting with tubulin heterodimers, leading to the disruption of the microtubular cytoskeleton. Full article
Show Figures

Figure 1

15 pages, 4175 KiB  
Article
High Temperature-Induced Spindle Destabilization Results in Aborted Pollen Production in Populus
by Zhiqun Li, Yifan Zhao, Xuetong Cheng, Bo Kong, Yaru Sang, Qing Zhou, Jian Wu and Pingdong Zhang
Forests 2023, 14(4), 797; https://doi.org/10.3390/f14040797 - 13 Apr 2023
Cited by 1 | Viewed by 1610
Abstract
High temperature can induce the production of 2n gametes and aborted pollen during microsporogenesis in Populus canescens. However, the mechanism by which high temperature induces pollen abortion remains unknown. Here, pollen abortion was induced by exposing male flower buds of P. canescens [...] Read more.
High temperature can induce the production of 2n gametes and aborted pollen during microsporogenesis in Populus canescens. However, the mechanism by which high temperature induces pollen abortion remains unknown. Here, pollen abortion was induced by exposing male flower buds of P. canescens to 38 and 41 °C; pollen morphology, meiotic abnormalities, defects of the meiotic microtubular cytoskeleton, and tapetum development were characterized. We found that the dominant meiotic stage, temperature, and duration of treatment significantly affected the percentage of high temperature-induced aborted pollen. Damaged spindle microtubules and depolymerized microtubular cytoskeletons were observed, which resulted in many lagging chromosomes at anaphase I and II as well as aneuploid male gametes and micronuclei, thereby generating aborted pollen grains. Tapetum disintegration was also delayed. The anther dehisced normally, and some viable pollen grains were released. However, no sufficient evidence showed that the delayed degradation of the tapetum was responsible for pollen abortion. Full article
(This article belongs to the Section Genetics and Molecular Biology)
Show Figures

Figure 1

Back to TopTop